Several biological indicators correlate with irAEs during the occurrence of abnormal immune responses. Although none of the biomarkers have been definitively validated to predict irAEs, some may have clinical applications. Several biomarkers emerge after the onset of irAEs; they get evaluated after treatment. In addition to their prospective clinical utility, these biomarkers confirm the mechanism by which irAEs occur. Below we will review several classes of biomarkers and discuss their relations with the aforementioned ICIs-related toxicity (Fig. 2).
T cell-related biomarkers in TME
T cells represent essential targets for ICIs. Some evidence suggests that they play roles in irAEs through decreased clonality, increased TCR diversity, and defective Treg function. Moreover, immune infiltrating cells in TME can also affect T cells' function by secreting related proteins (Fig. 2a).
One mouse model of paraneoplastic neurological disease found that antigen-specific T cells migrated into the cerebellum and CD8+ T cells caused significant neuroinflammation and paraneoplastic neurological disease by killing Purkinje neurons after anti-CTLA4 treatment [45]. In this study, T-cell clones were detected by their presence in peripheral blood and possibly by next-generation sequencing (NGS) to provide early indications of irAEs. Investigators carried out single-cell RNA sequencing on biopsy tissue samples from colitis; they reported a significant accumulation of CD8+ T cells that share TCR sequences with tissue-resident memory cells [46]. This suggests that changes in TCR diversity are associated with GI toxicity. The pathogenesis of ICIs-induced nodular disease is unclear, but it is noteworthy that CTLA4 expression decreases in Tregs in patients with nodular disease after the application of immune checkpoint blockade, causing defective Treg suppression. In comparison, no specific serum manifestations have been found for ICIs-induced nodular disease [47, 48].
The bivariate regression model of T cell-activated lymphocyte cytosolic protein 1 (LCP1) and ADP-dependent glucokinase (ADPGK) combined pharmacovigilance data and omics data to assess the association between multi-omics factors and odds of reporting irAEs in different cancer types, suggesting that combination of them has the best predictive for irAEs accuracy [49]. However, the forecasting value of LCP1 in diverse tumors is unclear and still needs further validation. CD163 is a receptor expressed by anti-inflammatory (M2) macrophages. It mainly locates in TME. M2 macrophages are characterized by their immunosuppressive properties, thus correlating with poor prognosis. In autoimmune diseases, as well as melanoma patients during anti-PD1 therapy [50], soluble CD163 (sCD163) obtained by proteolytic shedding of the receptor arose. A study showed that the density of CD163+ M2 macrophages was associated with inadequate recovery of immune-related renal function. In addition, researchers found an abnormity in the absolute rate of change concerning CD163 in serum: a significant increase in patients with irAEs compared with those without symptoms [51]. The soluble CTLA-4 isoform (sCTLA-4) engages in membrane-bound isoform inhibition [52] and blocks the interaction between full-length CTLA-4 and Tregs and B7 ligands expressed by self-reactive T cells, thereby enhancing T cell cytotoxicity and reducing Treg cells' immunosuppressive function, known as increased T cell activation. Analysis of sera from IPI-treated MM patients by ELISA revealed that higher baseline sCTLA-4 levels were associated with any irAEs (P = 0.029), particularly in the digestive tract [53].
Hence, the abnormal subset of T cell clones found in inflammatory tissues and blood circulation suggests that detecting activated T cells in peripheral blood may be an early indicator of the occurrence of irAEs. Second, the relationship between detecting immune-infiltrating lymphocyte-associated proteins activated in TME and different types of immunotoxicity still needs further investigation. Ultimately, the concentration of soluble biomarkers in the peripheral circulation of the organism exerts essential roles in predicting the occurrence of irAEs and verifying their involvement in the mechanism of T-cell-induced irAEs.
B cell biomarkers
Despite predictive biomarkers of ICB and strategies to enhance clinical response focused on the T-cell compartment, early B-cell changes are proposed to predict irAEs after anti-PD-1 and anti-CTLA-4 combination therapy [54]. The presence of tissue-specific autoantibodies after treatment and high baseline autoantibody levels detected prior to treatment are associated with the subsequent occurrence of irAEs, which supports the idea that some mechanisms of immunotherapy-related toxicity are mediated by B cells (Fig. 2b).
Researchers analyzed immune cells from a small peripheral blood sample. High proportions of CD19+ B cells link to grade ≥ 3 (OR = 0.063, P = 0.02) and grade ≥ 2 (P = 0.051) irAEs, although the latter showed only a significant trend. In addition, a prospective study showed that patients with a more than 2-fold increase in CD21lo B cells and plasma cells tended to develop irAEs [54]. This minor finding may indicate CD21lo B cells as a promising source of toxicity-associated autoantibodies.
Previous studies have identified many auto-specific antibodies in various organ tissues. ICIs imbalance the body's immune system, producing specific antibodies that recognize and bind platelets, such as αIIbβ3 and GP1b antibodies, which continuously remove platelets from peripheral blood by activating the monocyte-macrophage system in the body [55, 56]. For example, nivolumab induced and increased the production of platelet-specific IgG antibodies, which could destroy platelets and lead to immune-related thrombocytopenia, in patients with non-small cell lung cancer (NSCLC). One mouse model purified a cardiac-specific 30 kDa protein from heart extracts and identified it as cardiac troponin I (cTnI). Mice lacking the PD-1 immunosuppressive coreceptor developed autoimmune dilated cardiomyopathy and produced high titers of autoantibodies against a cardiac-specific 30 kDa protein.
Defined baseline high levels or abnormal increases in autoantibodies correspond to tissue-specific irAEs. Several studies have already detected specific autoantibodies in patients with endocrine irAEs. Among them, patients with pituitary dysfunction show increased titers of anti-guanine nucleotide-binding protein G (olf) subunit alpha and anti-integrin 2B antibodies [57], and anti-pituitary antibodies (APAs) can predict the presence of pituitary autoimmunity measured by indirect immunofluorescence [38]. It is unclear whether APAs are positive at baseline or before the onset of pituitary dysfunction. Therefore, we need further studies to clarify the utility of APAs as biomarkers of pituitary irAEs. Interestingly, anti-GAD, anti-IA2, and anti-zinc transporter 8 antibodies were positive prior to initiation of ICI therapy in a patient with ICI-induced Type 1 diabetes mellitus (T1DM) [42], suggesting these autoantibodies being biomarkers that suggest the presence of T1DM in a subset of patients at baseline. Implicated antigenic targets for bullous pemphigoid (BP) include the hemidesmosomal structural proteins of the dermoepidermal junction, BP180 (collagen XVII), and BP230 [58]. Serological detection revealed elevated antibodies against BP180 and BP230. Another study [59] measured serum levels of BP180, BP230, and type VII collagen in patients receiving PD-1/PD-L1 inhibitor treatment at baseline and 8 weeks post-treatment. The results showed that increased autoantibody production against hemidesmosomal protein BP180 IgG was significantly correlated with skin irAEs only at baseline, but not with BP230 and type VII collagen.
In general, B cell-related biomarkers are mainly precursor B cells and autoantibodies. While using autoantibodies to predict irAEs offers the edge of application: pre- and post-treatment sera of ICIs are readily available, and the relationship between their baseline levels and immunotoxicity in various organ tissues can be easily verified.
Antigen-related biomarkers
The same antigenic epitopes in tumor tissues and other organs can enhance their recognition through cross-presentation, so common antigen overexpression indicates the occurrence of irAEs. Besides, the original antigens may be recognized as non-self substances and cross-react after systemic application of ICIs. Their specific antigenic determinant clusters require further exploration as risk factors (Fig. 2c).
One study performed NGS using myocardial T cells and tumor T cells from patients who developed fatal myocarditis, they identified high levels of muscle-specific antigens (desmin and troponin) in the tumors [60], suggesting that T cells probably target antigens shared by melanoma, skeletal muscle, and heart. It is hypothesized that immune-mediated myocarditis results from the over-activation of adaptive immune responses that target common epitopes of myocardial and tumor cells. Typically, PD-L1 and PD-L2 are expressed on sundry types of self-cells (tubular epithelial cells, endothelial cells, fibrillary reticulocytes, pancreatic islet cells, astrocytes, and neurons), reducing autoimmune reactions and host organ injury [11]. In vitro, pretreating IFNg-stimulated HK-2 cells with anti-PD-L1 significantly enhanced co-cultured and mitogen-activated Jurkat or IL-2 secreted from human peripheral blood T cells [61]. PD-L1 overexpression serves as a central mechanism for inducing immunosuppression; T cells undergo apoptosis and overexpression, impairing vascular permeability in other tissues and organs. Therefore, we hypothesized that PD-L1 overexpression may be a potential biomarker to predict the occurrence of irAEs.
Some patients underwent a mild cross-reactivity between tumor antigens and dorsal root ganglia (DRG) after immunotherapy. Pembrolizumab upregulates cytotoxic T-cell responses to DRG via activating the slaughter of cancer cell cycle and subacutes sensory impairment [62]. It is reasonable to hypothesize that an intrinsic kidney antigen, initially tolerated but recognized as non-self with the brake of CTLA-4/PD-1 signaling in self-reactive T cells, could be responsible for acute tubulointerstitial nephritis [63, 64].
Cytokine-related biomarkers
Cytokines are small secreted proteins. (Fig. 2d) As means of intercellular communication, they act locally and distantly to influence cellular responses and cause intercellular coordination. ICIs may tilt the balance of this environment toward inflammation and autoimmunity [65].
Chemokines, a group of low molecular weight peptides, induce chemotaxis of different leukocyte subtypes and function primarily to recruit leukocytes to sites of inflammation and play a role in tumor growth, angiogenesis and organ sclerosis. Endocrine epithelial cells secrete CXCL9, CXCL10, and CXCL11 after IFN-γ stimulation, which in turn migrate CXCR3-expressing type 1 T helper lymphocytes and emit IFN-γ. Thereby they mediate autoimmune inflammation sustaining in endocrine glands [66]. The proportion of circulating CXCR4+ T cells and CCR7+ B and T cells (but not CXCR5) in peripheral blood mononuclear cells (PBMC: lymphocytes + monocytes) is significantly reduced in patients with autoimmune thyroid disease. This systemic effect of active intrathyroidal lymphoid tissue is a possible novel marker of thyroid autoimmune disease activity. The B-cell sequestrant CXCL13 mainly exists in mature B cells, CD4+ follicular Th cells, and activated Treg cells and takes part in autoimmune, inflammatory diseases [67].
Aside from organ specificity, cytokine levels may be associated with the onset and severity of irAEs. Inflammation has an integral role in the mechanisms. The specific milieu of circulating cytokines promotes and reflects changes in systemic immune responses, including anti-inflammatory cytokines (IL-10, TGF-β), pro-inflammatory innate immune cytokines (IL1, IL-6), and pro-inflammatory adaptive immune cytokines (IFN-γ, IL-17) [68]. The central pro-inflammatory cytokine IL-6 in plasma is commonly elevated in inflammatory arthritis after ICI treatment. Baseline serum IL-6 levels regressed negatively with the incidence of irAEs in melanoma patients treated with ipilimumab [69]. Surprisingly, an increase in IL-6 levels after ICI treatment was relevant to irAEs. In patients presenting with immune-associated refractory esophageal strictures, a pathogenic pro-inflammatory CD4+ T cell subpopulation expressing IL-17a (c-Kit-CD161+ MDR1+ Th17 cells) serves a vital role in the refractory autoimmune response [70], and IL-6 induces the maturation of naïve CD4+ T cells into Th17 cells. As a result, the IL-6-Th17 pathway may function in the pathogenesis of irAEs. Effective treatment of colitis with tocilizumab or infliximab [69] has also been shown to correlate with high levels of cytokines IL-6 or TNF, respectively. Moreover, elevated baseline serum IL-17 levels were similarly associated with an increased risk of diarrhea and severe colitis. Thus, these studies suggest IL-6 and IL-17 as predictive markers for developing irAEs. One study divided 26 patients with advanced malignant diseases treated with ICIs into two groups: those who presented with thyroid irAE and those who did not. Higher levels of serum IL-1β, IL-2, and GM-CSF at baseline were found when measured before and at 4 weeks of treatment, while early decreases in IL-8, G-CSF, and MCP-1 were significantly associated with thyroid irAEs [71].
Cytokines have diversity, posing challenges for assessing the clinical relevance of irAEs. Consequently, we warrant prospective studies involving larger cohorts to determine the clinical utility of cytokine analysis tools.
Blood cell biomarkers
Blood cell count is a routine clinical test. It has received extensive attention as a highly available specimen and a signal for irAEs, and the primary focus has been on leukocytes, neutrophils, lymphocytes, and eosinophils (Fig. 2e).
Multivariate logistic regression analyses showed that pre-treatment neutrophil-to-lymphocyte ratio (NLR) < 4.3 was significantly correlated with the risk of grade 3–4 irAEs, while the rate of change in NLR after treatment, ΔNLR > 120% were remarkably associated with increased risk [72], suggesting NLR a valid prognostic and morbidity in grade 3–4 irAEs marker. This may be because of the balance among circulating neutrophils and lymphocytes, which reflects anti-tumor forces. In turn, dysregulation of this balance might ascribe to the pathogenesis of irAEs. In a clinical trial, patients treated with ipilimumab showed increased levels of neutrophil activation markers CD177 and CEACAM1 and a higher risk of gastrointestinal irAEs [73]. The possible mechanism is that activated neutrophils spread inflammation to organs, subsequently causing tissue damage; On the other hand, they control the activation of T and B cells through antigen expression and cytokines. However, a colonoscopy in patients using ipilimumab for 1–2 weeks revealed focal neutrophilic cryptitis, neutrophilic infiltration, excess plasma cells in lamina propria, and lymphocytic cryptitis increased 3 weeks before the appearance of symptoms of colitis (grade 2 or higher diarrhea) [74]. Researchers also concluded that these findings lack reliability as biomarkers for predicting immune-mediated colitis in the clinical setting [74]. As such, activated neutrophils are controversial biomarkers for predicting ICB-associated intestinal irAEs.
Recently, researchers provided evidence that ICIs act on lung tissue after activation of Th2 cells, and serology shows eosinophil formation [75]. A high peripheral eosinophil count is relevant to the incidence of gastrointestinal irAEs and a generalized maculopapular rash [76, 77]. One mouse model used a combination of CTLA-4 blocking antibody 9D9 and/or indoleamine 2,3-deoxygenase 1 (IDO1) inhibitor epacadostat (mimic the effects of PD1 blockade) in wild-type and PD-1(-/-) mice [78]. This synergistically induced immune-associated liver injury and immune cell infiltration and found sites of hepatocyte necrosis frequently surrounded by mononuclear immune cells. ICIs increased the expression of genes pertaining to hepatocyte death, leukocyte migration, and T cell activation in the liver, and synergistic action of anti-PD-1 and CTLA-4 with IDO1 triggered this process.
Peripheral blood markers, including lymphocyte and eosinophil counts and neutrophil-to-lymphocyte ratios [79], have led to more practical clinical applications because of their invasiveness and ease of routine measurement, but are not widely used to predict the occurrence of irAEs due to their poor specificity. Notably, the accuracy and reliability of blood counts as predictors of irAEs still need further weighing and validation.
Genomic biomarkers
HLA is a highly polymorphic complex consisting of closely linked gene seats (Fig. 2f). It contains abundant alleles, and the variants are unique and may correlate with the occurrence of certain irAEs. [80] There is a significant association between the human leukocyte antigen HLA-DQB1*03:01 and colitis in malignant melanoma patients treated with nivolumab. Researchers compared the HLA allele frequencies of 11 cancer patients who developed endocrine dysfunction after ICB treatment with the Japanese HLA haplotype database and found that HLA-DR15, B52, and Cw12 may be susceptibility factors for pituitary irAEs (induced secondary adrenal insufficiency) [81]. One small sample study suggested a predominance of HLA-DR4 in patients with ICI-induced autoimmune T1DM [42]. A higher frequency of HLA-DQB1*06:01, DPB1*09:01, and DRB5*01:02 was reported in another study [82]. Therefore, the HLA haplotype can be used as a predictive marker for pituitary irAE. A clinical trial enrolled patients with advanced cancer treated with ICIs. Those with a genetic variant of human leukocyte antigen, HLA-DRB1*11:01, tended to develop pruritus [80]. Two advanced cancer patients carrying HLA class II (HLA-II) germline alleles HLA-DRB1*15:01 and HLA-DQB1*06:02 susceptible to systemic lupus erythematosus (SLE) may trigger immune-related SLE [83].
In short, genetic markers suggest early irAEs, and the association between their specific SNPs and specific tissues remains to be investigated.
Autoimmune disease
In order to avoid disease flares in patients with advanced cancer who have autoimmune diseases of their own, we generally exclude the use of immunotherapy in such individuals when conducting studies of clinical ICIs. However, the association between a history of autoimmune disease and irAEs has been confirmed in some experimental models or patients without treatment according to the protocol. A study [84] established a mouse experimental autoimmune myocarditis model by intraperitoneal injection of α-myocardial heavy chain (MyHC-α) fragments into mice. They administered anti-PD-1 antibodies (mPD1ab) to mice at two-time points, simultaneously with and after MyHC-a fragment administration, and observed that ICI-induced autoimmune myocarditis may be related to the activity of the autoimmune system prior to ICI administration. Subsequent administration of mPD1ab significantly increased infiltration of a cluster of differentiation (CD)4 and F4/80-positive cells compared with those administered concomitantly. This suggests that we should try to avoid the use of ICIs in patients with autoimmune diseases. Patients with preexisting AIDs are at an increased risk of irAEs in advanced cancer patients receiving anti-PD-1 immunotherapy compared to those without. Thus, experience with autoimmune disease is a high-risk factor for predicting irAEs.